Hemangiosarcoma is a rare, highly aggressive cancer that originates from endothelial cells lining blood vessels, forming malignant tumors that can rapidly metastasize to distant organs.[1] In humans, it is often referred to as angiosarcoma and accounts for approximately 1-2% of all soft tissue sarcomas (with an incidence of about 1-2 cases per million people annually), typically presenting as high-grade malignancies with a poor prognosis.[2][3] While uncommon in humans, hemangiosarcoma is one of the most prevalent cancers in dogs (accounting for 5-7% of all canine malignancies), particularly affecting older, large-breed animals such as German Shepherds, Golden Retrievers, and Labrador Retrievers, where it frequently involves the spleen or heart.[4][5]In humans, angiosarcomas most commonly arise in the skin (especially the head, neck, or scalp), breast, liver, or heart, and are associated with risk factors including prior radiation therapy, exposure to chemicals like vinyl chloride, chronic lymphedema (as in Stewart-Treves syndrome), and certain genetic conditions such as neurofibromatosis type 1.[2] Symptoms often include bruise-like lesions on the skin that may ulcerate, painless swelling, or organ-specific effects like abdominal pain or shortness of breath depending on the site; early detection is challenging due to its nonspecific presentation and aggressive nature, leading to a 5-year survival rate of around 35% even with treatment.[6][2] Diagnosis typically involves biopsy with immunohistochemical staining for markers like CD31 and CD34, supported by imaging such as CT or MRI to assess spread.[2]In dogs, hemangiosarcoma is characterized by its tendency to cause internal bleeding upon tumor rupture, resulting in sudden collapse, pale gums, rapid breathing, or even death; subcutaneous forms may appear as red or bruised masses on the skin.[4] The disease is diagnosed through abdominal ultrasound, chest X-rays, or cytology of fluid from effusions, with confirmation via histopathology.[4] Treatment usually combines surgical removal (e.g., splenectomy) with chemotherapy using agents like doxorubicin, though median survival remains short at 4-6 months for visceral forms, highlighting the need for ongoing research into targeted therapies shared between human and canine models due to their biological similarities.[4][7]
General Overview
Definition and Classification
Hemangiosarcoma is a rare and aggressive malignant neoplasm originating from endothelial cells that line blood vessels and lymphatic vessels.[2] In humans, it is more commonly termed angiosarcoma, while the designation hemangiosarcoma is frequently used in veterinary medicine, particularly for cases in dogs and cats.[8] This tumor is characterized by its rapid proliferation, local invasion, and high propensity for metastasis, distinguishing it from benign vascular lesions.[9]As a subtype of soft tissue sarcoma, hemangiosarcoma falls under the broader category of vascular tumors in oncological classifications.[10] The World Health Organization (WHO) Classification of Soft Tissue and Bone Tumours (2020 edition) categorizes angiosarcoma (hemangiosarcoma) as a malignant vascular tumor, emphasizing its endothelial differentiation through morphological and immunophenotypic features.[10] Histologically, it exhibits variants including epithelioid (with rounded cells mimicking carcinoma), spindled (elongated cells forming slit-like spaces), and well-differentiated forms (resembling normal vascular channels with anastomosing sinusoids).[2] These variants are identified via immunohistochemistry markers such as CD31, CD34, and factor VIII-related antigen, which confirm endothelial origin.[9]Hemangiosarcoma must be differentiated from benign hemangioma, a non-malignant vascular proliferation that lacks invasive growth, atypia, and metastatic potential.[2] While hemangiomas form well-circumscribed, encapsulated lesions with regular endothelial cells, hemangiosarcomas show pleomorphic, hyperchromatic nuclei, mitotic activity, and destructive infiltration into surrounding tissues.[8] In veterinary contexts, such as canine cases, histological grading further classifies hemangiosarcomas into low, intermediate, and high grades based on criteria like cellular atypia and necrosis, aiding in prognosis assessment.[11] Cutaneous forms are often more indolent and amenable to surgical excision compared to visceral variants, which arise in internal organs like the spleen or heart.[8]
Epidemiology
Hemangiosarcoma, also known as angiosarcoma in humans, is a rare malignancy accounting for 1-2% of all soft tissue sarcomas.[12] Its annual incidence in the United States is approximately 0.3 per 100,000 person-years, with over 1,300 new cases diagnosed in 2019 alone.[13] In humans, the disease predominantly affects older adults, with a medianage at diagnosis of 71 years and a peak incidence in the 60- to 80-year-old age group; there is a slight female predominance, with 54.5% of cases occurring in women.[13] Global incidence remains low at around 0.1-0.3 per 100,000 across populations.[14]In veterinary medicine, hemangiosarcoma is far more prevalent in dogs than in other species, representing up to 7% of all canine malignancies and serving as the most common malignant splenic tumor.[15] The annual incidence risk in dogs is approximately 0.036% across all ages, rising to 0.074% in those aged 5 years and older.[16]Canine cases typically occur in middle-aged to older dogs, with a median age of 10 years, and show a male predominance, particularly for visceral forms; breeds such as German Shepherds, Golden Retrievers, Labrador Retrievers, and Flat-Coated Retrievers are overrepresented.[16][17]Hemangiosarcoma is rare in cats, comprising less than 1.5-2% of nonhematopoietic neoplasms, with only sporadic diagnoses in necropsy series (e.g., 18 cases in 3,145 feline necropsies over 11 years).[18]Feline cases most commonly affect middle-aged to older cats, with a mean age of 10 years and no clear breed or gender predisposition beyond a slight male skew in reported series.[18]In horses, hemangiosarcoma is sporadic and uncommon, with a reported prevalence of 0-0.7% in examined horses, primarily manifesting as cutaneous forms.[19] Equine cases lack strong demographic trends but are noted in adult animals without specific age, gender, or breed overrepresentation in limited reports.[19]
Pathogenesis
Etiology and Causes
Hemangiosarcoma primarily arises from the neoplastic transformation of endothelial cells that line blood vessels, leading to aggressive vascular tumors. In dogs, the cellular origin is increasingly understood to involve hematopoietic precursors or bone marrow-derived progenitors that migrate to vascular niches and differentiate into endothelial-like cells, rather than solely mature endothelial cells. This origin is supported by studies showing hemangiosarcoma cells expressing markers of both endothelial and hematopoietic lineages, such as CD31 and CD34.[20][8]Genetic mutations are central to the initiation of hemangiosarcoma, particularly in canine cases where somatic alterations in key tumor suppressor and oncogene pathways drive uncontrolled proliferation. Activating mutations in PIK3CA occur in approximately 45% of splenic hemangiosarcomas, often at hotspot residue 1047, leading to hyperactivation of the PI3K/AKT/mTOR signaling pathway. Inactivating mutations in TP53 are found in about 35% of cases, typically involving the DNA-binding domain and resulting in loss of tumor suppression. PTEN mutations, affecting around 10% of tumors, further dysregulate the PI3K pathway by removing inhibitory phosphate groups from PIP3. These genetic changes collectively promote endothelial cell survival and angiogenesis, initiating tumorigenesis.[12][17]In humans, where the tumor is termed angiosarcoma, certain environmental carcinogens have been established as triggers for hepatic forms, including exposure to thorotrast (a historical radiocontrast agent), arsenic (from medicinal or industrial sources), and vinyl chloride (an industrial chemical). These agents induce sinusoidal cell hyperplasia and reticulin excess as precursors to malignant transformation, with cases showing similar histopathological progression to idiopathic ones. Despite these identified triggers, the etiology of hemangiosarcoma remains idiopathic in the majority of cases across species, with no single initiating factor discernible in most instances.[21][17]Rare viral associations have been noted in cats, where hemangiosarcoma can emerge alongside other tumors induced by feline sarcoma virus (FeSV), a recombinant retrovirus derived from feline leukemia virus (FeLV) that requires FeLV for replication and promotes poorly differentiated sarcomas in young cats.[22]
Risk Factors
Hemangiosarcoma, also known as angiosarcoma in humans, is associated with several non-modifiable and modifiable risk factors that elevate susceptibility, particularly in specific subtypes. Prior radiation therapy, often administered for breast cancer or other malignancies, significantly increases the risk of developing angiosarcoma, with tumors frequently arising in irradiated fields years later.[6] Chronic lymphedema, especially post-mastectomy, can lead to Stewart-Treves syndrome, a rare lymphangiosarcoma variant occurring in approximately 0.45% of affected patients, characterized by angiosarcomatous changes in edematous tissues.[23] Chemical exposures, such as vinyl chloride in industrial settings, are strongly linked to hepatic angiosarcoma, with historical cohorts of exposed workers showing elevated incidence rates.[24]Ultraviolet (UV) radiation exposure is a key risk for cutaneous forms, particularly on sun-exposed areas like the head and neck.[25] In humans, while no strong hereditary patterns exist for the majority of cases, rare familial clusters have been reported in association with genetic syndromes including neurofibromatosis type 1, Li-Fraumeni syndrome, Maffucci syndrome, Klippel-Trenaunay syndrome, and germline BRCA1/2 mutations.[26][27]In dogs, hemangiosarcoma exhibits clear genetic predispositions, with larger breeds such as Golden Retrievers, German Shepherds, and Labrador Retrievers facing substantially higher risks due to polygenic inheritance identified through genome-wide association studies.[28] For cutaneous variants, UV exposure plays a role, especially in dogs with lightly pigmented or short-coated skin, increasing susceptibility to dermal tumors on exposed areas.[4] Advanced age is a universal non-modifiable factor across species, with most cases in dogs occurring in middle-aged to older individuals, typically over 6 years.[29]Among other animals, horses with light-colored or unpigmented skin, such as Haflingers or Appaloosas, are predisposed to dermal hemangiosarcoma due to chronic UV exposure, which damages endothelial cells in sun-exposed regions like the periorbital area or limbs.[30]Age remains a consistent risk factor in veterinary species, including cats and horses, where tumors predominantly affect mature animals. No definitive protective factors against hemangiosarcoma have been established in humans or animals, though minimizing modifiable exposures like UV radiation or chemicals may mitigate risk in susceptible populations.[26]
Pathophysiology
Tumor Biology
Hemangiosarcoma is a malignant tumor originating from endothelial cells lining blood vessels, characterized histologically by pleomorphic, immature endothelial cells that form irregular, slit-like vascular spaces often filled with blood and prone to hemorrhage and necrosis.[8] These tumors exhibit varied morphologies, including capillary, cavernous, or solid patterns, with the neoplastic cells lining malformed vascular channels and expressing endothelial markers such as CD31 and von Willebrand factor.[31] The hemorrhage-prone nature arises from the fragile, disorganized vascular structures, leading to frequent intratumoral bleeding and necrosis.[8]The tumor's aggressive angiogenesis drives the formation of tortuous, dilated vascular channels filled with blood, promoting rapid growth and instability.[8] This process involves upregulation of pro-angiogenic factors, resulting in chaotic vessel formation that supports the tumor's nutrient demands but contributes to its fragility.[32] At the molecular level, hemangiosarcoma is driven by recurrent somatic mutations, including TP53 (in >70% of canine cases and frequent in humanangiosarcoma), PIK3CA, and NRAS in dogs, and TP53, POT1, and ATRX in humans, which disrupt cell cycle control, activate oncogenic signaling, and promote genomic instability.[7][26] Dysregulation of vascular endothelial growth factor (VEGF) signaling plays a central role, with hemangiosarcoma cells producing high levels of VEGF to stimulate autocrine and paracrine angiogenesis.[33] Genetic studies in mouse models demonstrate that even partial loss of endothelial-derived VEGF significantly reduces tumor incidence, highlighting its essential role in initiation and progression.[33] Hypoxia-inducible factor 1-alpha (HIF-1α) further contributes by responding to the hypoxic tumor environment, enhancing VEGF expression and other genes that promote vascularization and survival under low-oxygen conditions.[8]Due to its vascular origin, hemangiosarcoma exhibits high metastatic potential through early hematogenous spread, allowing tumor cells to disseminate via the bloodstream.[17] Common metastatic sites include the lungs and liver, where emboli of neoplastic endothelial cells lodge and form secondary tumors, often present at diagnosis in over 60% of cases.[17] This rapid dissemination is facilitated by the tumor's ability to invade vessel walls and exploit the circulatory system.[34]The tumor microenvironment in hemangiosarcoma is enriched with inflammatory cells and factors that support growth and enable immune evasion.[35] Proinflammatory cytokines such as interleukin-8 (IL-8) and chemokines like CXCL12 recruit immune cells, creating a permissive niche that enhances tumor cell migration, invasion, and survival while polarizing macrophages toward an immunosuppressive M2 phenotype.[8] Regulatory T cells (Tregs) accumulate in the microenvironment, suppressing antitumor immunity through checkpoint molecules like CTLA-4, thereby facilitating progression and metastasis.[35] This inflammatory milieu also intersects with angiogenic pathways, amplifying the tumor's aggressive behavior.[32]
Common Sites of Origin
Hemangiosarcoma, a malignant tumor arising from vascular endothelial cells, most commonly originates in visceral organs in dogs, with the spleen accounting for 50-65% of cases.[36] The right atrium of the heart represents another primary visceral site in dogs, often leading to pericardial effusion and acute clinical deterioration due to its propensity for rupture.[4] In humans, cardiac angiosarcoma similarly favors the right atrium, comprising a significant portion of rare primary heart tumors.[27]The liver serves as a frequent visceral site, particularly in humans following exposure to chemicals such as vinyl chloride, where hepatic angiosarcoma develops in association with industrial toxins.[9] In dogs, liver involvement often occurs as a primary tumor or metastasis, though less dominant than splenic origins. Cutaneous and subcutaneous forms arise in sun-exposed skin areas, linked to ultravioletradiation; these account for approximately 13% of hemangiosarcomas in dogs, typically in lightly pigmented breeds.[37] In humans, cutaneous angiosarcoma predominates, affecting about 60% of cases, most often on the head and neck in elderly individuals.[9]Less common sites include the retroperitoneum, bone (such as the radius), and brain, where tumors exhibit similar vascular origins but rarer incidence across species.[8] Multicentric disease, involving simultaneous primaries at multiple sites like spleen and heart, complicates staging and occurs in up to 25% of canine cases, reflecting the tumor's propensity for widespread endothelial involvement.[5] Cardiac tumors demonstrate heightened aggressiveness, frequently causing life-threatening hemorrhagic events compared to more indolent cutaneous variants.[38]
Clinical Presentation
Signs and Symptoms
Hemangiosarcoma presents with nonspecific clinical signs that vary by species, tumor location, and whether it is cutaneous or visceral. Due to its vascular origin, the tumor can cause hemorrhage, leading to anemia, weakness, lethargy, pallor of mucous membranes, anorexia, weight loss, and exercise intolerance.[2][17]In visceral forms, particularly common in dogs, acute manifestations often result from tumor rupture, causing sudden collapse, abdominal distension due to hemoperitoneum (especially splenic origin), and hypovolemic shock.[17] Cardiac involvement may lead to arrhythmias, pericardial effusion with tamponade, dyspnea, and syncope.[17] Metastatic spread can worsen these through additional hemorrhage or organ dysfunction.[17]Cutaneous forms, more prevalent in humans and horses, typically manifest as bruise-like lesions, red to purple nodules, or raised masses on the skin that may ulcerate and bleed intermittently, resembling hematomas.[6] In humans, visceral sites like breast or liver may cause localized pain, swelling, fatigue, or weight loss.[2] Systemic effects such as cachexia and coagulopathies can occur across forms.[17]Early-stage disease is often asymptomatic and found incidentally during examinations or imaging.[17]
Species Variations
Hemangiosarcoma, also known as angiosarcoma in humans, exhibits variations in clinical presentation across species, influenced by predominant tumor locations.In humans, it most frequently manifests as cutaneous lesions on the scalp, face, or other sun-exposed areas, presenting as bruise-like purple or red nodules, plaques, or ulcers that enlarge gradually and may bleed intermittently.[6] Visceral presentations in the breast, liver, or spleen are less common and associated with risk factors like radiation or chemicals, causing localized pain, swelling, or systemic symptoms like fatigue and weight loss.[3][27]In dogs, it predominantly affects visceral organs, with the majority of cases originating in the spleen or right atrium of the heart, leading to rapid metastasis and high risk of rupture causing acute internal hemorrhage.[4] Signs include sudden collapse, profound weakness, pale mucous membranes, abdominal distension, and rapid breathing.[39] Cutaneous forms are less common and generally have a better prognosis.[40]In cats, common sites include cutaneous areas and the spleen, with presentations such as skin masses, lethargy, anorexia, weight loss, vomiting, or abdominal swelling.[41] The disease has high metastatic potential and poor prognosis, with risks of internal bleeding and collapse similar to dogs.[42]In horses, it primarily arises in dermal or conjunctival sites, often in lightly pigmented areas due to chronic UV exposure, manifesting as raised red-to-black nodules or ulcerative lesions on the skin, eyelids, or nictitating membrane.[43] Signs are usually localized (e.g., ocular discharge, swelling, lameness), with visceral forms rare and causing nonspecific symptoms like weight loss or dyspnea.[44]Detection often occurs incidentally in animals during routine exams, while in humans, cutaneous or visceral symptoms prompt evaluation.[29][45]
Diagnosis
Diagnostic Methods
Diagnosis of hemangiosarcoma typically involves a combination of clinical evaluation, imaging, laboratory tests, and histopathological confirmation to identify the vascular origin of the tumor and assess its extent. In dogs, initial suspicion often arises from nonspecific signs like weakness or abdominal distension, prompting targeted diagnostics based on common sites such as the spleen or heart.[17] In humans (where termed angiosarcoma), presentation may include skin lesions or organ-specific symptoms, with diagnosis emphasizing biopsy and imaging for sites like skin, breast, or liver.[2]Imaging plays a central role in detecting primary tumors and metastases. In dogs, abdominal ultrasound is commonly used to identify splenic or hepatic masses, appearing as heterogeneous, hypoechoic lesions with cavitation or mixed echogenicity, and to detect peritoneal effusion.[17] For cardiac involvement, echocardiography reveals right atrial masses and associated pericardial effusion, aiding in preoperative planning.[17] Advanced cross-sectional imaging like computed tomography (CT) or magnetic resonance imaging (MRI) enhances staging by detecting pulmonary or distant metastases; CT shows heterogeneous contrast enhancement in vascular phases, while MRI delineates soft tissue invasion with intermediate T1 and hyperintense T2 signals.[17] In humans, similar imaging is used, with positron emission tomography (PET)-CT often employed for staging due to its sensitivity for metastases.[2]Laboratory findings often support the diagnosis through evidence of hemorrhage and coagulopathy. In dogs, regenerative or nonregenerative anemia is frequent due to internal bleeding, accompanied by thrombocytopenia in approximately 70% of cases, schistocytes, and acanthocytes on blood smears.[46] Coagulation panels may reveal disseminated intravascular coagulation (DIC) risks, including prolonged prothrombin time, activated partial thromboplastin time, hypofibrinogenemia, and elevated fibrin degradation products, reflecting microangiopathic damage.[47]Leukocytosis with a left shift can also occur.[17] In humans, lab findings are less uniformly hemorrhagic but may include anemia or elevated liver enzymes if hepatic involvement.[2]Cytological examination via fine-needle aspiration (FNA) is a presurgical screening tool but is often inconclusive due to hemodilution and hemorrhage, potentially mimicking hemangiomas. In humans, core biopsy is preferred over FNA for better sampling.[17] Definitive diagnosis requires histopathology from incisional or excisional biopsies, revealing infiltrative vascular channels lined by malignant endothelial cells with hemorrhage and necrosis.[17] Immunohistochemistry confirms endothelial origin using markers such as CD31 and Factor VIII-related antigen (von Willebrand factor), which highlight vascular structures and differentiate from other sarcomas; in humans, additional markers like ERG and Fli-1 may be used.[48][2]Staging employs adapted systems like the three-stage model (I: localized, II: ruptured or invading locally, III: metastatic), incorporating tumor size, lymph node involvement, and distant spread assessed via imaging and sometimes bone marrow evaluation.[17] This guides prognosis and therapy, with complete staging including thoracic radiographs or CT for pulmonary assessment.[17] Human staging follows sarcoma guidelines, often using TNM classification with PET-CT for M stage.[2]Advanced techniques include flow cytometry to detect endothelial progenitor markers like CD34, CD133, and c-kit in circulating cells, potentially identifying early disease or primitive origins in research settings, though not routine clinically.[49]Contrast-enhanced ultrasound (CEUS) and elastography further characterize lesions by vascular patterns and stiffness, with shear wave velocities over 2.6 m/s suggesting malignancy.[17]
Differential Diagnosis
Hemangiosarcoma, particularly in its visceral forms affecting the spleen, heart, or liver, can mimic several other conditions due to shared features such as hemorrhage, vascular involvement, or mass effects. In dogs, the most common differentials for a splenic mass include benign vascular lesions like hemangioma, which is distinguished by its lack of metastasis, well-circumscribed borders on imaging, and absence of cellular atypia on histopathology.[50] Hematomas, non-neoplastic collections of blood often resulting from trauma or coagulopathies, present similarly with hemoabdomen but lack neoplastic cells on cytology or biopsy and typically resolve without progression.[50] Lymphoma, a hematologic malignancy, may involve the spleen diffusely or as nodules and is differentiated by lymphoid markers such as CD3 or CD79a on immunohistochemistry, contrasting with the endothelial markers (e.g., CD31, CD34) positive in hemangiosarcoma.[51]Other malignant tumors, such as leiomyosarcoma originating from smooth muscle, can be distinguished histologically by positive staining for smooth muscle actin and desmin, while melanoma often shows pigmentation and S-100 protein expression.[50] Non-neoplastic mimics like infectious abscesses are identified through cytology revealing inflammatory cells, bacteria, or fungi, often accompanied by systemic signs such as fever, and coagulopathies (e.g., disseminated intravascular coagulation) may cause spontaneous bleeding without a discrete mass, confirmed by coagulation profiles showing prolonged prothrombin time or thrombocytopenia.[50] In humans, where the condition is termed angiosarcoma, key differentials include benign hemangiomas (lacking infiltrative growth and atypia), Kaposi sarcoma (associated with HHV-8 and positive for that viral marker), and epithelioid hemangioendothelioma (intermediate grade with less pleomorphism), primarily differentiated via histopathology and immunohistochemistry for vascular endothelial markers.[2]Distinction from these mimics relies on imaging characteristics, such as vascular flow voids on Doppler ultrasonography or contrast enhancement patterns indicating hypervascularity in hemangiosarcoma, alongside biopsyhistology demonstrating malignant endothelial cells lining irregular vascular spaces.[50] The absence of atypia, mitotic figures, or invasion in benign entities like hemangioma further aids differentiation.[2] However, acute hemorrhage frequently obscures tumor features in both species, complicating preoperative assessment and often necessitating splenectomy for definitive diagnosis in dogs, as fine-needle aspiration yields low sensitivity due to sampling errors and bleeding risks.[50]
Treatment
Surgical Options
Surgical intervention serves as the primary treatment modality for hemangiosarcoma, aiming to achieve complete tumor resection when feasible, though its aggressive nature and vascularity often limit curative outcomes.[52] For splenic hemangiosarcoma, particularly prevalent in dogs, splenectomy is the standard procedure, involving complete removal of the spleen to address the tumor and prevent life-threatening rupture.[17] This approach is often palliative in cases of tumor rupture, stabilizing the patient by halting acute hemorrhage and allowing time for further management.[4]Cardiac hemangiosarcoma presents significant surgical challenges due to the tumor's location within or near the heart, frequently requiring adjunctive pericardectomy to relieve pericardial effusion and prevent cardiac tamponade.[53] Tumor resection in the right atrium or other cardiac sites may be attempted under cardiopulmonary bypass, but such procedures are technically demanding and carry high risks; in humans, they are rarely curative given the tumor's propensity for early metastasis.[54] For cutaneous hemangiosarcoma, wide local excision with 1-2 cm lateral margins and deep extension to the fascial plane offers the potential for cure if clean margins are achieved and no distant spread is present.[17]In cases of unresectable hemangiosarcoma, such as advanced hepatic or multifocal disease, debulking surgery may be performed to reduce tumor burden and alleviate symptoms like vascular obstruction or organ compression, though it does not address microscopic disease.[55] Common complications across these procedures include intraoperative hemorrhage from the tumor's fragile vascular structure and postoperative cardiac arrhythmias, particularly following splenectomy or pericardial manipulation.[56][40] For incomplete resections, adjuvant therapies are often recommended to target residual disease.[52]
Adjuvant Therapies
Adjuvant therapies for hemangiosarcoma aim to complement surgical intervention by targeting microscopic disease, reducing recurrence, or providing palliation when surgery is incomplete or not feasible. In veterinary medicine, particularly for canine cases, doxorubicin-based chemotherapy protocols are the cornerstone of adjuvant treatment following splenectomy for splenic hemangiosarcoma. Single-agent doxorubicin administered every 2-3 weeks for 4-5 cycles has been shown to extend median survival from approximately 1-3 months with surgery alone to 5-7 months. Combination protocols, such as vincristine, doxorubicin, and cyclophosphamide (VAC), offer similar benefits without significantly improving outcomes over single-agent therapy but may be used in cases of advanced disease. In human angiosarcoma, which shares histological similarities with hemangiosarcoma, adjuvant doxorubicin-based regimens are also employed post-resection, though evidence is limited due to rarity, with response rates around 20-30% in metastatic settings.Radiation therapy serves primarily as a palliative modality for hemangiosarcoma, particularly in cutaneous, subcutaneous, or unresectable visceral tumors where surgical margins are incomplete. In dogs with nonsplenic hemangiosarcoma, palliative protocols delivering 20-30 Gy in 3-5 fractions achieve subjective tumor reduction in up to 70% of cases, with median survival times of about 3 months and minimal acute toxicity due to the vascular nature limiting curative intent. For cardiac hemangiosarcoma in dogs, hypofractionated radiation (e.g., 12 Gy in a single fraction) has demonstrated feasibility and short-term stabilization in pilot studies, though long-term control remains challenging owing to the tumor's radiosensitivity and metastatic propensity. In humans, stereotactic body radiotherapy is occasionally used adjuvantly for localized angiosarcoma, providing local control rates of 50-70% in small series, but systemic progression often predominates.Targeted therapies, particularly anti-angiogenic agents, represent an emerging adjuvant strategy exploiting hemangiosarcoma's vascular origin. Tyrosine kinase inhibitors like pazopanib, which inhibit VEGF and PDGF receptors, have shown promise in human angiosarcoma as second-line therapy, with phase II trials reporting disease control rates of 50% at 3 months, especially in cutaneous subtypes, and median progression-free survival of 3-4 months. In preclinical canine models, similar multi-kinase inhibitors demonstrate anti-angiogenic effects, though clinical adoption remains investigational. These agents are typically reserved for cases refractory to chemotherapy, with monitoring for hypertension and hepatotoxicity essential.Immunotherapy approaches are largely experimental for hemangiosarcoma, focusing on vaccines and checkpoint inhibitors to harness antitumor immunity. In canine trials, vaccines such as the extracellular vimentin (eVim)-targeted vaccine combined with doxorubicin post-surgery have improved outcomes, with a 2025 phase I/II study showing a median survival of 235 days and 1-year survival rate of 44% in dogs with visceral hemangiosarcoma, compared to 136 days and 14% with doxorubicin alone, without added toxicity. Other approaches, including the EVM peptide vaccine, have reported 1-year survival rates of 35.7% in adjuvant settings. For human angiosarcoma, checkpoint inhibitors like pembrolizumab are under evaluation in trials, yielding objective responses in 20-40% of vascular sarcomas with high mutational burden. Ongoing veterinary trials, including multivalent vaccines for hemangiosarcoma, aim to validate these as adjuncts to standard care.Supportive therapies are integral to adjuvant management, addressing complications like anemia and hemorrhage from tumor rupture. In dogs, whole blood or packed red blood cell transfusions are commonly required pre- or post-surgery for severe anemia (hematocrit <20%), stabilizing patients and enabling chemotherapy tolerance, with transfusion rates up to 50% in splenic cases. Pain management with opioids or NSAIDs, alongside fluid therapy, enhances quality of life during multimodal treatment. In humans, similar supportive measures, including erythropoietin-stimulating agents for chemotherapy-induced anemia, support adjuvant regimens without altering core outcomes.
Prognosis
Survival Rates
Hemangiosarcoma, also known as angiosarcoma in humans, exhibits highly variable survival outcomes depending on the primary site and disease stage. In humans, localized cutaneous forms have reported 5-year survival rates of approximately 30-50%, reflecting better outcomes when tumors are confined to the skin and amenable to complete surgical resection.[52] In contrast, metastatic visceral hemangiosarcomas, often involving organs such as the liver or spleen, carry a dismal prognosis with 5-year survival rates of approximately 10-20% and median overall survival of 6-16 months, even with multimodal therapy.[52][57]In dogs, the most common species affected, untreated hemangiosarcoma typically results in median survival times of 1-3 months due to rapid progression and rupture-related complications. Surgical intervention alone, such as splenectomy for splenic tumors, extends median survival to 4-6 months, while adjunctive chemotherapy (e.g., doxorubicin-based protocols) can achieve up to 6-12 months in select cases, though 1-year survival rates are approximately 10-20% in reported cohorts. Cutaneous variants generally confer longer survival, with median times exceeding 2 years for early-stage, actinic forms following excision.[17]For cats, survival is more favorable in cutaneous hemangiosarcoma compared to other forms, where complete surgical excision can yield median survival times of approximately 9-12 months overall, with longer outcomes possible for superficial lesions based on studies through 2023. Visceral forms, however, are aggressive, with median survival of approximately 2-3 months post-surgery and limited extension from chemotherapy.[18][58]In horses, isolated cutaneous or ocular hemangiosarcomas respond well to surgical excision, often resulting in cure rates exceeding 80% and long-term survival (e.g., over 2 years in small cohorts), provided margins are clean and no dissemination occurs. Disseminated visceral disease, though rarer, portends a poor outcome similar to other species.[43]Across species, survival is strongly influenced by the stage at diagnosis—early detection markedly improves prognosis—and the anatomic site, with cutaneous presentations consistently outperforming visceral ones due to lower metastatic potential. Prognosis data are based on studies up to 2023, with no major changes reported as of 2025.[17]
Prognostic Factors
Prognostic factors for hemangiosarcoma encompass a range of tumor, patient, and biological characteristics that influence disease progression and outcomes, with variations observed across species but particularly well-studied in dogs.[17] Tumor stage at diagnosis is a primary determinant, where localized disease (stage I) confers a more favorable course compared to metastatic involvement (stage III), as the presence of distant spread significantly shortens survival due to widespread dissemination often detected in over 75% of cases at necropsy.[17] Similarly, the anatomic site of the tumor plays a critical role; cutaneous hemangiosarcomas generally exhibit better prognoses owing to their superficial nature and lower metastatic potential, whereas visceral sites such as cardiac or splenic locations are associated with aggressive behavior and poorer outcomes, largely attributable to the high risk of rupture and internal hemorrhage.[17][59]Histological grade further refines prognosis, with high-grade tumors demonstrating more rapid progression and reduced survival relative to low-grade variants, reflecting increased cellular atypia and mitotic activity that drive invasion and metastasis.[17] Among patient-related factors, age shows inconsistent impact in dogs, typically affecting middle-aged to geriatric individuals without a strong independent prognostic effect, though advanced age beyond 70 years correlates with worse outcomes in human cases.[17][60] Performance status, including the presence of anemia or thrombocytopenia, adversely affects prognosis in visceral forms, signaling systemic compromise and advanced disease burden.[17]Effusion, particularly pericardial in cardiac hemangiosarcoma, indicates advanced involvement and is linked to diminished survival, as it often accompanies metastasis and hemodynamic instability.[17] Arrhythmias, commonly seen in cardiac presentations, similarly portend a guarded outlook by reflecting tumor-induced myocardial disruption and complicating clinical management.[17]Response markers provide additional prognostic insight through histopathological evaluation. The Ki-67 proliferation index, a measure of cellular replication, is elevated in aggressive hemangiosarcomas and serves as an independent predictor of shorter survival, with indices exceeding 56% in splenic canine cases associated with poorer outcomes.[61][62] Vascular invasion identified on biopsy heightens the risk of metastasis, particularly in subcutaneous or dermal tumors, thereby worsening prognosis by facilitating early systemic spread.[17] These factors collectively guide clinical decision-making, emphasizing the need for thorough staging and histopathological assessment to tailor expectations.[17]
Occurrence in Humans
Incidence and Demographics
Hemangiosarcoma, also known as angiosarcoma in humans, is a rare malignancy accounting for approximately 1-2% of all soft tissue sarcomas, with an estimated annual incidence in the United States of about 1,300 cases as of 2019.[3][63] The age-adjusted incidence rate has been reported at around 3 cases per 1,000,000 person-years, reflecting a notable increase from earlier decades.[63]Demographically, angiosarcoma predominantly affects older adults, with a median age at diagnosis of 71 years and the majority of cases occurring in individuals aged 60 years or older.[64] It shows a slight female predominance overall (approximately 54.5% of cases), though this varies by subtype; for instance, cutaneous forms exhibit a male-to-female ratio of about 2:1, while breast angiosarcomas are nearly exclusive to women.[64][65] Non-Hispanic White individuals represent the largest racial group affected, comprising roughly 75% of cases in population-based analyses.[66]In terms of site distribution, cutaneous angiosarcomas are the most common, accounting for about 60% of cases and frequently involving the head and neck region.[52] Other primary sites include deep soft tissues (approximately 36%), breast (13-22%), visceral organs such as the liver and heart (17%), and bone (3%).[66][67]The incidence of angiosarcoma has risen over time, doubling in the US from 657 cases in 2001 to 1,312 in 2019, with an annual increase in the age-adjusted rate of 1.6%.[63] This trend is partly attributed to a growing proportion of secondary cases linked to prior radiation therapy for other cancers, particularly breast cancer.[68] Globally, angiosarcoma remains rare with an incidence of 1-3 cases per million population annually, but rates appear higher in industrialized regions due to associations with occupational chemical exposures, such as vinyl chloride in liver angiosarcomas.[66][69]
Human-Specific Management
Management of angiosarcoma in humans requires a multidisciplinary approach, typically involving sarcoma reference centers where dedicated teams of pathologists, radiologists, surgeons, medical oncologists, and radiation oncologists collaborate to tailor treatment plans.[70] This coordinated care is essential due to the tumor's aggressive nature and variable presentations, ensuring comprehensive evaluation and individualized strategies.[71]For large or locally advanced tumors, neoadjuvant chemotherapy is often employed to shrink the lesion, facilitating subsequent surgical resection and improving margin status.[55] Regimens commonly include anthracyclines or taxanes, administered prior to surgery to enhance resectability in cases where upfront excision might otherwise be challenging.[72]In advanced or metastatic disease, targeted therapies play a key role; weekly paclitaxel has emerged as a standard option, demonstrating response rates around 40-50% in pretreated patients.[73] For subsets expressing PD-L1 or with microsatellite instability-high features, anti-PD-1 inhibitors such as pembrolizumab have shown durable responses in select cases, particularly within clinical trial settings.[74]Radiation therapy is integral, often delivered postoperatively to address close or positive margins, reducing local recurrence risk in resectable cases.[75] For inoperable tumors, definitive radiation serves as a primary modality, sometimes combined with chemotherapy to achieve local control.[52]Enrollment in clinical trials is strongly encouraged, especially for novel anti-angiogenic agents like bevacizumab, which targets vascular endothelial growth factor and has demonstrated efficacy in metastatic angiosarcoma with response rates up to 25% in phase II studies.[76]Palliative care focuses on symptom management and quality of life in metastatic settings, where systemic therapies aim to prolong survival; median overall survival in this context ranges from 8 to 12 months with current approaches.[77]
Occurrence in Dogs
Prevalence and Breed Predispositions
Hemangiosarcoma accounts for approximately 5-7% of all tumors diagnosed in dogs, making it one of the most common malignancies encountered in veterinary practice.[38] It represents about 5% of all canine cancers overall and is frequently identified during necropsy, occurring in 0.3% to 2% of examined cases.[5] While exact incidence rates vary by study, population-based analyses indicate an annual incidence of around 0.035% (95% CI: 0.03-0.038%) in dogs aged 5 years or older in the United Kingdom as of 2019.[16]Certain dog breeds exhibit significantly elevated risks for developing hemangiosarcoma, highlighting a genetic predisposition particularly in purebred populations. German Shepherds face the highest risk, with an odds ratio of 6.14 (95% CI: 4.73–7.98) compared to reference breeds.[16] Golden Retrievers and Labrador Retrievers also show increased susceptibility, with odds ratios of 2.05 (95% CI: 1.33–3.17) and 1.42 (95% CI: 1.11–1.83), respectively.[16] This heritability is evident in pedigree dogs, where breed-specific genetic factors contribute to higher incidence rates, though environmental influences may play a minor role.[38] Geographic variations in prevalence appear minimal, with similar patterns observed across North America, Europe, and other regions without strong regional disparities.[16]The condition predominantly affects older dogs, with a median age at diagnosis of 9 to 10 years, though cases can occur in younger animals.[78] There is a slight male bias, with neutered males showing a modestly higher risk (odds ratio 1.49, 95% CI: 1.17–1.90) compared to entire females.[16] Among diagnosed cases, visceral forms dominate, comprising the majority—estimated at over 70%—and most commonly involving the spleen or heart at the time of presentation.[79]
Canine-Specific Clinical Features
Hemangiosarcoma in dogs often presents with acute and life-threatening clinical features due to its vascular nature, distinguishing it from more indolent tumors. A significant risk is sudden death, with studies reporting that 25% to 65% of cases involve splenic rupture leading to massive internal hemorrhage and hypovolemic shock.[80][81] This rupture can occur without prior warning, causing rapid collapse and fatality if untreated.[82]When involving the heart, particularly the right atrium, hemangiosarcoma frequently causes pericardial effusion, leading to specific cardiac signs such as syncope and muffled heart sounds on auscultation.[83] These symptoms arise from cardiac tamponade, where accumulated blood in the pericardial sac compresses the heart, impairing cardiac output and resulting in episodes of fainting, especially during exertion.[84]Cutaneous hemangiosarcoma in dogs typically manifests as rapidly growing, fragile lesions on the skin, often in hypopigmented or lightly haired areas, which may reach 5-10 cm in diameter within weeks to months.[85] These tumors appear as red to black, raised masses that can ulcerate and bleed easily due to their vascular composition.[17]Early metastasis is a hallmark, with approximately 80% of dogs harboring micrometastases at the time of diagnosis, frequently remaining occult and undetected by routine imaging.[86] This widespread dissemination contributes to the aggressive course, even in apparently localized disease.Behavioral changes in affected dogs often include acute weakness following exercise, accompanied by pale mucous membranes indicative of ongoing hemorrhage.[87] These signs reflect the tumor's propensity for vascular leakage, leading to anemia and reduced oxygen delivery to tissues.[88]
Occurrence in Other Animals
In Cats
Hemangiosarcoma is a rare malignancy in cats, accounting for less than 1.5% to 2% of all nonhematopoietic neoplasms reported in feline pathology registries.[18] It most commonly arises in the skin (cutaneous or dermal form) or viscera, particularly the spleen, with the overall incidence in cats being substantially lower than in dogs.[18] Unlike in canines, no strong breed predispositions have been identified, though the condition predominantly affects older cats with a mean age of approximately 10 years (range 4–19 years).[18]Cutaneous hemangiosarcoma in cats typically presents as indolent, non-ulcerative nodules on the head, pinnae, paws, or trunk, often discovered incidentally during grooming or examination.[18] In contrast, visceral forms are more aggressive and manifest with systemic signs such as anorexia, lethargy, splenomegaly, collapse, or respiratory distress due to internal hemorrhage or organ involvement, commonly affecting the spleen, liver, or intestines.[89]Metastasis occurs less frequently in cats compared to dogs, with evidence of spread in only about 38% of staged cases, primarily to the lungs or liver.[18]Prognosis varies significantly by location and treatment. Cutaneous lesions often respond well to wide surgical excision, particularly when complete margins are achieved, potentially offering a curative outcome with median survival exceeding 283 days and no tumor-related deaths in some cohorts.[18] Visceral hemangiosarcoma, however, carries a grave outlook, with many cats euthanized shortly after diagnosis due to severe clinical signs and anemia; for those undergoing splenectomy or supportive care, median survival is around 77 days.[89] Adjuvant therapies like chemotherapy are infrequently used but may extend survival in select cases.[18]
In Horses
Hemangiosarcoma is an uncommon neoplasm in horses, primarily presenting as cutaneous or conjunctival tumors arising from vascular endothelial cells in sun-exposed areas. These tumors most frequently affect the third eyelid, bulbar conjunctiva, and lightly pigmented skin, such as the ventral abdomen, in horses with pale or dilute coat colors living in sunny climates. Chronic ultraviolet light exposure is a key risk factor, often associated with solar elastosis in affected tissues.[43][90][91]Internal hemangiosarcomas are rare in horses and typically involve the spleen or skeletal muscle, manifesting clinically as acute colic from splenic rupture and hemorrhage or lameness due to intramuscular masses. The disease predominantly occurs in middle-aged to older horses, with reported mean ages ranging from 12 to 16 years across cases, and no sex bias has been identified. Thoroughbreds may be overrepresented, though no definitive breed predisposition exists.[92][43][91]Cutaneous and conjunctival forms often appear as slow-growing, exophytic, hyperemic vascular masses, with ulceration possible in advanced skin lesions; metastasis is uncommon in these localized presentations. In contrast, internal cases frequently disseminate rapidly to multiple organs, including the lungs, heart, and kidneys, leading to systemic signs like dyspnea, epistaxis, or subcutaneous swelling.[43][92]Surgical excision, including conjunctivectomy, keratectomy, or enucleation for ocular involvement, is the primary treatment for localized tumors and is often curative, with many horses surviving over 2 years post-surgery without recurrence. Adjunctive therapies like cryotherapy may be used, but their efficacy is not well-established. For disseminated internal disease, prognosis is guarded to poor, with supportive care such as fluid therapy and transfusions providing only palliative relief, as euthanasia is common within weeks of diagnosis.[43][91][92]